• Non ci sono risultati.

The safety of monoclonal antibodies in asthma

N/A
N/A
Protected

Academic year: 2021

Condividi "The safety of monoclonal antibodies in asthma"

Copied!
12
0
0

Testo completo

(1)

Workflow: Annotated pdf, CrossRef and tracked changes

PROOF COVER SHEET

Journal acronym: IEDS

Author(s): Giovanni Passalacqua, Andrea Matucci, Alessandra Vultaggio, Diego Bagnasco, Marcello Mincarini, Enrico Maggi and Giorgio Walter Canonica

Article title: The safety of monoclonal antibodies in asthma Article no: 1186641

Enclosures: 1) Query sheet 2) Article proofs Dear Author,

1. Please check these proofs carefully. It is the responsibility of the corresponding author to check these and approve or amend them. A second proof is not normally provided. Taylor & Francis cannot be held responsible for uncorrected errors, even if introduced during the production process. Once your corrections have been added to the article, it will be considered ready for publication.

Please limit changes at this stage to the correction of errors. You should not make trivial changes, improve prose style, add new material, or delete existing material at this stage. You may be charged if your corrections are excessive (we would not expect corrections to exceed 30 changes).

For detailed guidance on how to check your proofs, please paste this address into a new browser window: http://journalauthors.tandf.co.uk/production/checkingproofs.asp

Your PDF proof file has been enabled so that you can comment on the proof directly using Adobe Acrobat. If you wish to do this, please save the file to your hard disk first. For further information on marking corrections using Acrobat, please paste this address into a new browser window: http://journalauthors.tandf.co.uk/production/acrobat.asp

2. Please review the table of contributors below and confirm that the first and last names are structured correctly and that the authors are listed in the correct order of contribution. This check is to ensure that your name will appear correctly online and when the article is indexed.

Sequence Prefix Given name(s) Surname Suffix

1 Giovanni Passalacqua 2 Andrea Matucci 3 Alessandra Vultaggio 4 Diego Bagnasco 5 Marcello Mincarini 6 Enrico Maggi

(2)

Queries are marked in the margins of the proofs, and you can also click the hyperlinks below.

Content changes made during copy-editing are shown as tracked changes. Inserted text is inred fontand revisions have a red indicator©. Changes can also be viewed using the list comments function. To correct the proofs, you should insert or delete text following the instructions below, but do not add comments to the existing tracked changes.

AUTHOR QUERIES

General points:

1. Permissions: You have warranted that you have secured the necessary written permission from the appropriate copyright owner for the reproduction of any text, illustration, or other material in your article. Please see http://journalauthors.tandf.co.uk/permissions/usingThirdPartyMaterial.asp.

2. Third-party content: If there is third-party content in your article, please check that the rightsholder details for re-use are shown correctly.

3. Affiliation: The corresponding author is responsible for ensuring that address and email details are correct for all the co-authors. Affiliations given in the article should be the affiliation at the time the research was conducted. Please see http://journalauthors.tandf.co.uk/preparation/writing.asp.

4. Funding: Was your research for this article funded by a funding agency? If so, please insert‘This work was supported by <insert the name of the funding agency in full>’, followed by the grant number in square brackets ‘[grant number xxxx]’.

5. Supplemental data and underlying research materials: Do you wish to include the location of the underlying research materials (e.g. data, samples or models) for your article? If so, please insert this sentence before the reference section:‘The underlying research materials for this article can be accessed at <full link> / description of location [author to complete]’. If your article includes supplemental data, the link will also be provided in this paragraph. See <http://journalauthors.tandf.co.uk/preparation/multimedia.asp> for further explanation of supplemental data and underlying research materials.

6. The CrossRef database (www.crossref.org/) has been used to validate the references. Changes resulting from mismatches are tracked inredfont.

AQ1 Please check whether the ORCID for Matucci Andrea is correct. AQ2 Please confirm whether the details added to the affiliation are correct. AQ3 Please confirm whether the corresponding author details are correct.

AQ4 Please confirm whether the presentation of term“name of the molecule [AND] safety [OR] side effects” is OK. AQ5 We have deleted figure citation (Figure 1) from the head level and inserted in the following paragraph. Please

check.

AQ6 Please define“GM-CSF” at first occurrence.

AQ7 Please confirm whether the edit in the sentence‘In some case reports, cases of omalizumab-related AEs on the pathogenic mechanism were sustained . . .’ conveys the intended meaning and amend if necessary.

AQ8 Please define“FEV1” at first ocurrence.

AQ9 The PubMed (http://www.ncbi.nlm.nih.gov/pubmed) and CrossRef (www.crossref.org/) databases have been used to validate the references. Mismatches between the original manuscript and PubMed or CrossRef are tracked in red font. Please provide a revision if the change is incorrect. Do not comment on correct changes. AQ10 Please provide missing volume number for the“Singh et al., 2011;” references list entry.

AQ11 Please provide missing volume number for the“Normansell et al., 2014;1” references list entry. AQ12 Please provide missing volume number/page number for the “Sakkal et al., 2016” references list entry.

(3)

How to make corrections to your proofs using Adobe Acrobat/Reader

Taylor & Francis offers you a choice of options to help you make corrections to your proofs. Your PDF prooffile has been enabled so that you can mark up the proof directly using Adobe Acrobat/Reader. This is the simplest and best way for you to ensure that your corrections will be incorporated. If you wish to do this, please follow these instructions:

1. Save thefile to your hard disk.

2. Check which version of Adobe Acrobat/Reader you have on your computer. You can do this by clicking on the“Help” tab, and then“About”.

If Adobe Reader is not installed, you can get the latest version free from http://get.adobe.com/reader/.

3. If you have Adobe Acrobat/Reader 10 or a later version, click on the“Comment” link at the right-hand side to view the Comments pane.

4. You can then select any text and mark it up for deletion or replacement, or insert new text as needed. Please note that these will clearly be displayed in the Comments pane and secondary annotation is not needed to draw attention to your corrections. If you need to include new sections of text, it is also possible to add a comment to the proofs. To do this, use the Sticky Note tool in the task bar. Please also see our FAQs here: http://journalauthors.tandf.co.uk/production/index.asp. 5. Make sure that you save thefile when you close the document before uploading it to CATS using the “Upload File” button on the online correction form. If you have more than one file, please zip them together and then upload the zip file.

If you prefer, you can make your corrections using the CATS online correction form. Troubleshooting

Acrobat help: http://helpx.adobe.com/acrobat.html Reader help: http://helpx.adobe.com/reader.html

Please note that full user guides for earlier versions of these programs are available from the Adobe Help pages by clicking on the link“Previous versions” under the “Help and tutorials” heading from the relevant link above. Commenting functionality is available from Adobe Reader 8.0 onwards and from Adobe Acrobat 7.0 onwards.

Firefox users: Firefox’s inbuilt PDF Viewer is set to the default; please see the following for instructions on how to use this and download the PDF to your hard drive:

(4)

REVIEW

The safety of monoclonal antibodies in asthma

Giovanni Passalacquaa, Andrea Matucci b, Alessandra Vultaggiob, Diego Bagnascoa, Marcello Mincarinia, Enrico Maggicand Giorgio Walter Canonicaa

a

5 Allergy and Respiratory Diseases, IRCCS San Martino,IST,University of Genoa,Genova,Italy

AQ2 ;bImmunoallergology Unit, AOU Careggi,University of

Florence,Florence,Italy;cCentre of Excellence DENOTHE, Department of Experimental and Clinical Medicine, University of Florence,Florence,Italy

ABSTRACT

Introduction: In the last two decades the knowledge of the mechanisms of the inflammatory processes underlying asthma rapidly evolved, several key mediators (cytokines and receptors) were identified, and 10 the laboratory techniques have allowed us to synthesize monoclonal antibodies highly specific for those target molecules. Nowadays, many biological agents are investigated in asthma (with anti IgE being the only commercially available). The clinical efficacy of some biologics was demonstrated in many cases, however, the safety issue has progressively emerged and has been recognized as a crucial aspect.

15 Areas covered: We summarized the currently available knowledge on the safety and side effects of biologics in asthma, as derived by reviews, meta analyses and clinical trials. PubMed was searched with the terms anti IL-x [AND] safety [OR] side effects, within the categories “clinical trial”, meta-analysis” and “review”. Case reports were excluded. The authors collegially selected the relevant entries to be included.

20 Expert opinion: Overall, the safety of most of the investigated agents seems to be satisfactory, a certain risk of side effects remains present, and is variable for the different molecules. Thus caution must be paid in evaluating the risk to benefit ratio. Specific biomarkers to predict the response to each biological are urgently needed to improve the safety profile.

ARTICLE HISTORY Received 22 March 2016 Accepted 29 April 2016 Published online xx xxx xxxx KEYWORDS

Asthma; allergic asthma; severe asthma; monoclonal antibody; cytokine; safety; adverse events

1. Introduction AQ1

25 Asthma, which remains a high-prevalence disease worldwide, [1,2] is currently recognized as a ‘heterogeneous disease’,[3] and this definition profoundly differs from what stated only few years ago in Guidelines, when asthma was considered as a single and uniform disease. This is the result of the more

30 detailed knowledge that we have gained on clinical presenta-tion, biological/immunological aspects, functional behavior,

and treatment approaches. Consequently, there have been many attempts to identify distinct phenotypes of asthma, according to clinical and biological criteria.[4] So far, a quite

35 clear distinction can be made only between the T-helper 2 (TH2) and low-TH2 phenotypes.[5] Within these large and partially overlapping groups, allergic asthma (AA) remains the paradigm of TH2-driven asthma, whereas other forms, such as adult-onset or smoke-related or obesity-related

40 asthma, well represent some of the usual low-TH2 presentations.

Starting from the well-known IgE-mediated mechanisms underlying allergic asthma, the pathophysiology of the disease was progressively and rapidly elucidated in thepasttwo

dec-45 ades, and alsoapplied to other forms of asthma. Cytokines, receptors, cell function, and chemokines were progressively identified and dissected. The evolving knowledge, in addition

to the evolving biotechnology, allowed to identify precise molecular targets to be addressed by specifically constructed

50

molecules to be applied in asthma, especially in the more severe forms.[6,7] This is the era of ‘precision medicine’,[8,9] where each single molecular target can be inhibited by biolo-gical agents (BA), monoclonal antibodies (MAbs) in particular. In less than 15 years, many MAbs targeted to single

compo-55

nents of the immune-mediated inflammation of asthma were synthesized, tested,and proposed for clinical use.[10] In par-allel to clinical use and large-population trials, the problem of the safety and tolerability of these approaches rapidly emerged: as any other drug, MAbs are not totally devoid of

60

side effects.[11,12]

The clinical efficacy, in the view of the personalized med-icine, remains the primary goal of MAb-based therapy but, due to the costs and affordability problems, the safety assumes a comparable importance. Here, we reviewed the

65

available data on the tolerability and safety profile of the MAbs currently used or under evaluation for the treatment of asthma (listed inTable 1). We searched PubMed for each of them using the terms name of the molecule [AND] safety

[OR] side effects, within the categories ‘clinical trial’, ‘meta- AQ4

70

analysis’ and ‘review’. Case reports were excluded. The authors collegially selected the relevant entries to be included and discussed.

CONTACTGiorgio Walter Canonica canonica@unige.it Allergy and Respiratory Diseases, Department of Internal Medicine, University of Genoa, Pad. Maragliano, Ospedale San Martino, L.go R. Benzi 10, 16133, Genova, Italy

AQ3

EXPERT OPINION ON DRUG SAFETY, 2016 http://dx.doi.org/10.1080/14740338.2016.1186641

© 2016 Informa UK Limited, trading as Taylor & Francis Group C/e: SP C/e QA: SR

(5)

2. An overview on the pathogenic mechanisms of asthma

AQ5

75 During thepasttwo decades, a great research effort has been undertaken to clarify the complex mechanisms,which regulate airways inflammation in asthmatic patients (Figure 1). The vast majority of our knowledge about the immunological aspects of asthma derives, for historical reasons, from the allergic model.

80 In such case,the triggering mechanism that starts the inflam-matory processes is well known: the specific IgE–allergen–mast cell interaction. It has become quite clear that the reaction of inhaled allergens with IgE-specific antibodies bound to FcεRI receptors on the surface of mast cells is not sufficient to

85 account for the persistent histological, pathophysiological,and clinical alterations that characterize the allergic asthma. The definition of phenotypic and functional activities of different T cell subsets based on their profiles of cytokine secretion has been of the primary importance. One type of T helper cells

90

(TH1) producesinterleukin (IL)-2 and IFN-g, whereas the other type (TH2) secretes IL-4, IL-5, IL-9,IL-13,and IL-10, but not IL-2 and IFN-g.[13,14] In particular, atopic subjects have a pre-exis-tent background that interacts with external factors (allergen exposure, maternal factors, infections, intestinal microbiota),

95

leading to an overproduction of IgE specific for ubiquitous and innocuous antigens. This abnormal production of IgE is the consequence of a relative imbalance between TH1 and TH2T cells. In fact, IL-4 and IL-13 favor IgE synthesis, whereas IL-5 increases eosinophil activation and survival.[15] IL-13, is

100

actively involved in inducing goblet cell metaplasia and bron-chial hyper-reactivity, and prime the upregulation of the adhe-sion molecule VCAM-1 and ICAM-1 on endothelial cells, that is a key step for eosinophils migration.[16] However, it should be remembered that T cells may play an important role also in

105

non-atopic asthma. In fact, similarly to what observed in atopic asthmatics, the biopsies taken from intrinsic asthmatics are characterized by large numbers of eosinophils and activated T lymphocytes.[17] Non-allergic asthmatics also display increased levels of IL-2 and IL-5, but not IL-4.[18]

Article highlights

● Asthma is a heterogeneous disease. Despite the clinical and func-tional aspects are common, different phenotypes/endotypes can be recognized. Allergic asthma is the most studied model.

● Several cytokines, cells and receptors are currently recognized as pivotal elements in asthma-related inflammation (IgE, IL-4, IL-5, IL13, IL-17)

● Monoclonal antibodies specific to relevant targets of inflammations are now available, and many of them are undergoing clinical trials. ● For anti IgE (in use since >10 years), there is a large amount of

evidence. Efficacy and safety data are available for many of the biological agents from the published clinical trials.

● Biologicals (in particular Monoclonal Antibodies) may exert both favourable and undesired side effects with various mechanisms. ● Non-specific adverse events (headache, local reactions, diarrhea etc.)

invariably resulted to be more frequent in active than in placebo groups.

● Overall, the biological agents tested in asthma displayed an overall efficacy to safety favourable profile, but this is not applicable at the same extent to each single agent.

● A‘precision medicine’ approach, including the search for biomarkers, would be necessary to appropriately prescribe biological agents, maintaining an optimal safety profile.

This box summarizes key points contained in the article.

APC DC ALLERGEN Th1

Th2

Eos

IL-5

B

IL-4 Y Y Y Y Y IgE MAST Fibro IL-13 MEDIATORS ALLERGEN CD4 Histamine LTs ADHESION MOLECULES LATE PHASE INFLAMMATION REMODELLING ADJUVANTS Neu CELL RECRUITMENT Th17 IL-17 IL-2 IL-13 TSLP

Figure 1.Schematic view of the allergic reaction. APC-DC: antigen presenting cell-dendritic cell; Eos: eosinophil: MAST: mastcell; Neu: neutrophil; TSLP: thymic stromal lymphopoietin.

Table 1.MAbs currently used or tested in asthma.

Name Function Structure

Phase status in asthma

Omalizumab Anti-IgE Humanized Commercialized (Xolair™)

Ligelizumab Anti-IgE Humanized IIb

Benralizumab Anti-interleukin (IL)-5 receptor Humanized II

Mepolizumab Anti-IL-5 Humanized III European Medicines Agencyand USFood and Drug Administration(FDA)approved (Nucala™)

Reslizumab Anti-Il-5 Humanized III FDA approved

Pascolizumab Anti-Il-4 Humanized II

Annukizumab Anti-Il-13 Humanized II

Lebrikizumab Anti-IL-13 Humanized III

Pitrakinra Anti-IL-4/IL-13 receptor Humanized IIb

Tralokinumab Anti-IL-13 Human IIb

Pitrakinra IL-4/IL-13 antagonist Mutein IIb(withdrawn)

Dupilumab Anti-IL-4/IL-13 Human IIb

Infliximab Anti-tumor necrosis factor-α (TNF-α) Chimeric III (withdrawn)

Golimumab Anti-TNF-α Human III (withdrawn)

Etanercept Soluble TNF-α receptor Fusion protein III (withdrawn)

Brodalumab Anti-IL-17 Human II

(6)

110 Eosinophils represent 1–6% of the circulating white blood cells. They are important effectors of the allergic inflammatory response, playing a primary role in the pathogenesis and severity of chronic inflammatory disorders of the airways.[19] The accumulation in the target tissues and the activation of

115 allergen-specific TH2-like cells as well as mast cells through an IgE-dependent pathway playa central role in orchestrating the airway allergic inflammation, by the recruitment of effector cells, mainly eosinophils. In this regard, different cytokines,

such as IL-3, IL-5, and GM-CSF

AQ6 are involved. IL-5 appears to

120 be the most specific for eosinophils by promoting their differ-entiation from bone marrow precursors, enhancing their adhe-sion to endothelial cells, prolonging their survival in target tissues and priming them for many activities as effector cells. [20–22] The recruitment of eosinophils to airways wall is

regu-125 lated by the production of chemokines,such as eotaxins 1, 2,

and 3 now called CCL11, CCL24,and CCL26, respectively.[23] In non-atopic‘intrinsic’ asthma, the accumulation and acti-vation of T cells able to produce cytokines, particularly IL-5, may be responsible for the peculiar eosinophilic airway

inflam-130 mation. Eosinophils can damage the respiratory mucosa by releasing basic proteins,oxygen-free radicals,and lipid med-iators, such as leukotriene C4 and platelet-activating factor, contributing to microvascular leakage, bronchoconstriction, mucus secretion, and shedding of the airway epithelium.

135 Eosinophils has been demonstrated to be alsoable to elabo-rate cytokines,such as IL-3, TGF-β, GM-CSF, IL-1, IL-6,and IL-5. [24] Recently, a key role in eosinophilic asthma has been attributed to the so called ‘type 2’ innate lymphoid cells (ILC2 cells), able to produce IL-5 as well as IL-9 and IL-13.[25]

140 ILC2 cells lack antigen-specific receptors, but like TH2 cells,

they react to the epithelium-derived cytokines IL-25, IL-33,and thymic stromal lymphopoietin (TSLP).[26]

3. Pathogenic mechanisms of adverse reaction to biological agents

145 The safety of BA is an important field of research consider-ing their growconsider-ing clinical applications. Adverse events (AEs) should be defined as any untoward medical occurrence associated with the use of a drug, whether or not consid-ered drug related (Table 2). The safety profile of biologicals

150 is negatively impacted by their immunogenicity, which leads to the production of specific anti-drug antibodies (ADA).[27] Similar to other biologicals, therapeutic MAbs are structurally immunogenic. They are classified as chimeric (variable regions from murine sources and constant regions

155 from human immunoglobulins) or humanized (containing

only the complementarity-determining regions of a murine immunoglobulin with the remaining being human), and fully human (Figure 2). These latter usually elicit minor, subclinical, and transient phenomena, but, sometimes, they

160

can induce complete cellular and humoral immune responses, which impact the efficacy. Various types of ADA were observed during biological treatments, mostly IgG, but also IgE, IgM,and IgA.[28] Patients developing high levels of ADA are more likely to show acute hypersensitivity

reac-165

tions, and both IgE and non IgE-mediated mechanisms may be involved. Other adverse events, such as serum sickness-like reactions appear to be associated with the presence of ADA, related to the formation of complement-binding immune complexes, with subsequent immune complexes

170

deposition, complement activation, and inflammatory infil-tration around small vessels. In fact, at immunofluorescence, the presence of complement deposition around vessels of skin specimens can be observed.[29] In addition, data from clinical trials and from real-life clinical practice suggested

175

that the currently used biologicals may constitute a risk factor for the reactivation of latent bacterial infections or parasitic infestations as in the case oftumor necrosis factor-α (TNF-α) blockers. In general, TH1 cells are more suitable for protection against intracellular infection agents, such as

180

Mycobacterium tuberculosis whereas the optimal reaction against parasites is provided by TH2 cells and IgE antibodies and eosinophils. Among biological agents used for the treatment of severe asthma and their association with the risk of infections, anti-IL-5 MAbs have been closely

followed-185

up. The role of T cells in the protection of mycobacterium is indirectly demonstrated by the effects of TNF-α blockers, also proposed for the treatment of severe forms of asthma. These biological agents are able to interfere with the

mycobacterium-specific-induced proliferation and cytokine

190

production by T cells, thus increasing the risk of infection or reactivation. TH1 cells produce IFN-γ, IL-2, and TNF-β, which activate the macrophages responsible for cell-mediated immunity to intracellular pathogens.[30]

Table 2.Adverse reactions to biological agents. Target-related events

● Infections

● Increased tumor susceptibility

● Autoimmunity (ADA, with possibly reduced efficacy of the MAb)

Drug-related events ● Infusion reactions

● Local (immediate or delayed) ● Systemic (immediate or delayed)

Chimeric MAbs

Humanized MAbs

Fully human MAbs

ADA Xenoantigens

Idiotypes Others (?)

Monoclonal antibodies: structure and immunogenicity

Figure 2.Schematic representation of therapeutic monoclonal antibodies and their structure-related immunogenic profile. ADA: anti-drugantibodies.

(7)

4. Anti-IgE (omalizumab)

195 For historical reasons, IgE was immediately regarded as an opti-mal target for a MAb-based strategy. In fact, IgEsare the primary trigger of the allergic reaction. The atopic subject produces abnormal amounts of allergen-specific IgE,which bind the high affinity receptor (FCεRI), widely distributed on tissue mastcells

200 and circulating basophils. The rationale of the anti-IgE strategy was to synthesize a MAb capable of specifically bind the heavy chain of the circulating IgE, thus preventing them from engaging with the receptor. The MAb obtained was a humanized one (human IgG with a 5% murine fraction), named omalizumab.

205 [31] Omalizumab underwent numerous phase II and phase III clinical trials in thepast15 years,[32] and was then approved for commercial use more than 10 years ago. Its indication is severely

uncontrolled asthma due to perennial allergens with a total IgE concentration of 30–1500kU/L and a body weight between 50

210 and 120 kg. Due to the long-standing clinical use,there is now a large amount of data available on its efficacy [33–35] and safety. Looking at the clinical trials so far published, the occurrence of AEs, including severe (SAEs) ones, was negligible and approxi-mated that of placebo groups.[36,37] In particular, in the

pub-215 lished trials the most frequently reported AEs were local (induration/irritation at the injection site) or systemic (headache, pharingytis,or rhinitis), none of them exceeding 10% of patients. Since the beginning, one of the major concerns was the risk of inducing or unmasking malignancies, since MAbs interferes

220 with the immune system. After many years of clinical use, it can be stated that the incidence of malignancies in the oma-lizumab-treated patients does not differ from that expected in the general population.[38,39] Another possible safety pro-blem is the occurrence of parasitosis, since IgE are well

225 known to be actively involved in the natural defensive response to parasites. According to the currently available data, an increased occurrence of parasite infestation during omalizumab treatment was not observed,[40] although a slight but not significant excess in geoelminthic infestations

230 (in at risk regions) was reported in a single study.[41] Finally, some cases of anaphylaxis, anaphylaxis-like reactions or serum sickness following omalizumab treatment have been sporadi-cally reported.[42,43] In some case reports,cases of omalizu-mab-related AEs on the pathogenic mechanism were 235 sustained by IgE antibodies

AQ7 directed against additives that are present in the drug formulation.[44] Indeed, according to the more recent survey conducted in the United States, the incidence of anaphylaxis or anaphylaxis-like events was about 0.09%, with 16 out of the 77 reported events occurring more

240 than 2 hafter the administration, thus leaving the diagnosis of anaphylaxis uncertain.[45] Based on these results, in the

United States, an observation period of at least 2 hand the prescription of autonjectable epinephrine are recommended, whereas those recommendations are not applied in most

245 other countries. When omalizumab was first commercialized and introduced in clinical practice, some reports of Churg– Strauss disease possibly related to the drug were published. These cases were attributed to the systemic steroid withdra-wal that unmasked a pre-existing disease. Nowadays, the

250 Churg–Strauss syndrome is suggested as a promising, although off-label, indication for omalizumab.[46]

In conclusion, looking back to more than 15 years of clinical trials and clinical use in real life, the safety profile of omalizu-mab is extremely satisfactory. Mild AEs (e.g. local) may occur,

255

but systemic and/or SAEs are very rare, if not anecdotal. From a very conservative and cautious point of view, a 2-hperiod of observation after each injection (and the prescription of auto-injectable epinephrine) are applied in theUnited States, but not in the remaining countries.

260 5. Anti-IL-5 and IL-5Ra chain (mepolizumab,

reslizumab, benralizumab)

IL-5 is secreted mainly by Th2 cells, mast cells, natural killer T cells, basophils, eosinophils, and type-2 ILCs. The IL-5R is a heterodimer composed ofβ-subunit, responsible for binding

265

of IL-5 and expressed both on progenitors of mature eosino-phils but also by basoeosino-phils, and the alpha subunit, necessary for signaling. IL-5 induces the maturation, activation, and recruitment of eosinophils.[47] The BA interfering with IL-5 and its receptor proposed for asthma are mepolizumab and

270

reslizumab (anti-IL-5 MAbs) and benralizumab (anti-IL-5Ra MAb).[24] Differently from the two anti-IL-5 MAbs, benralizu-mab targets IL-5Ra chain receptor and might thus also affect leukocytes via antibody-dependent cell-mediated cytotoxi-city.[48]

275

To date, few clinical trials with benralizumab have been completed, showing a significant efficacy in patients with

mild-to-severe asthma along with a reduction in peripheral blood and sputum eosinophils and eosinophil cationic protein. A single dose administered intravenously and subcutaneous

280

multipledose of benralizumab reduced eosinophil counts in airway mucosa and submucosa in sputum, and suppressed eosinophil counts in bone marrow and peripheral blood.[49– 51] Only nonspecific mild AEs (headache, fatigue) were described, and local reactions occurred in about 5% receiving

285

subcutaneous injections.

More data are available for mepolizumab, that invariably showed a significant benefit in eosinophilic asthma, character-ized by the decrease in the frequency of exacerbations,

increase in FEV1, improvement in quality of life, along with a AQ8

290

significant decrease in blood and sputum eosinophilia.[52–56] This was confirmed in a recent meta-analysis.[57] The most commonly reported AEs were nasopharyngitis, headache, chest pain, facial flushing, fatigue, upper respiratory tract infection in 10–25% of actively treated patients on average,

295

with few SAEs not related to treatment. Mepolizumab was usually given intravenously. In the case of subcutaneous administration, local side effects (induration, pain) occurred in about 10% patients (one case of anaphylaxis reported). Similar results were obtained with reslizumab in asthmatic

300

patients with sputum eosinophil levels of 3% and more. [58,59] In this case, there was not a significant difference in common AEs between placebo and active groups, but two

cases of anaphylaxis were reported with intravenous reslizumab.

305

As with anti-IgE, a possible safety problem is the occur-rence of parasitic infestation, since eosinophils are involved in the natural defense against parasites. Indeed, experiments in

(8)

parasite-infested mice and receiving anti IL-5 antibodies showed no change in immune response,[60] but no data are

310 available in humans. An indirect evidence of the safety of IL-5/ IL-5Ra blockers is that patients lacking eosinophils, do not display any abnormality related to eosinophil reduction.[61] Concerning the risk of malignancies, it is important to note the role of eosinophils in both anti-carcinogenic and

pro-carcino-315 genic effects depend on many factors mainly on the type of cancer. Eosinophil infiltration is considered unfavorable in Hodgkin’s lymphoma, but conversely, it was associated to a favorable prognosis in solid cancers (e.g. colorectal or pro-static). A variety of cytokines and factors produced by

eosino-320 phils such as major basic protein, eosinophil cationic protein, eosinophil-derived neurotoxin have either anti-tumor effects or stimulate tumor progression.[62] Although clinical experi-ence of IL-5 blockers in asthmatic patients is limited to clinical trials, additional information of the safety profile of these BA

325 were obtained by the data of patients with the hypereosino-philic syndromes treated with mepolizumab for several years: these subjects did develop no AE at all.[63]

6. Anti IL-4 and IL-13

Dupilumab antagonizes the effects of both IL-4 and IL-3 by

330 blocking the IL-4 receptor alpha chain. Wenzel and colleagues described the safety results in a clinical trial involving 104 asthmatics treated with dupilumab or placebo.[64] In this study, the overall proportion of AE in the two groups was similar (77% vs.81%). The reported AEs were mild or moderate

335 and only four patients had serious adverse event (threein the placebo group and one in the dupilumab group). There was no fatal event and most AEs were judged by investigators as not related to study drug. Six patients discontinued therapy: three in the placebo group (psoriasis, asthma exacerbation, 340 and upper respiratory tract infection) and three in the dupilu-mab group (worsening of bipolar disorder, angioedema and increase in asthma symptoms). The most common adverse events were injection-site reactions (29%), nausea, headache,

and nasopharingitis (8–13%),whichoccurred more frequently

345 with dupilumab. One case of angioedema, judged as treat-ment-related was reported. In addition, in 4/52 patients an unexpected increase in eosinophil count, not associated with asthma worsening was described.

The safety of pitrakinra, a mutein antagonizing both IL-4

350 and IL-13, was described by Wenzel and colleagues in a clinical trial in asthmatic people. Two group of patients, the first treated with pitrakinra by subcutaneous injections and the second one by inhalation route, were investigated in the same trial. The most frequent symptoms recorded were

355 about general disorders (headache, fatigue, musculoskeletal pain) and injection site reactions. Other reported adverse events were related to the nervous system (somnolence, dizzi-ness), respiratory,and thoracic disorders (wheezing, chest dis-comfort, dyspnea, and nasal obstruction) and gastro-enteric

360 symptoms (nausea, abdominal discomfort, and diarrhea). All those AEs were mild and occurred in about 10% of patients. [65] Despite the initial enthusiasm, no further trial with pitra-kinra was conducted after 2007.

The use of human anti IL-13 MAb lebrikizumab in asthma

365

was explored in few studies, with conflicting results. A pooled analysis of two phase II trials reported a significant reduction in asthma exacerbations, with a marginal effect on pulmonary function.[65] Corren et al.[66,67] showed a significant clinical efficacy of the treatment only in a subgroup of patients with

370

high blood periostin, whereas another trial failed to demon-strate a clinical efficacy.[68] The first study involved 219 adult patients with asthma. The overall occurrence of adverse events was similar in the active and placebo group, with the exception of musculoskeletal pain, more frequent in the active

375

group (13% vs. 5%). In the second ranging phase II (dose ranging) trial in mild asthma [68], the safety profile was good as well, with a similar proportion of AEs in placebo and active arms. Most of AEs were moderate, only one of the six SAEs in the lebrikizumab arm (Lofgren syndrome) was

consid-380

ered drugrelated. A third small study with lebrikizumab in 29 mild asthmatic patients, evidenced 61 AEs, with a greater incidence in the placebo group. Only one event (decreased platelet count) was judged as drug related. No patient was withdrawn because of AEs.[69]

385

Another anti IL-13 drug, tralokinumab, was evaluated in 194 asthmatic patients in a phase II study, at three different dose 150, 300,or 600 mg. Thesixreported SAEs were equally distributed between groups and not treatment related. The most frequent AEs in the Tralokinumab arm were asthma,

390

headache, nasopharyngitis, and local reactions. Interestingly, urinary tract infections were reported in 4% of the active-group patients as well as an increased eosinophil count in 2–6% of patients at week 13.[70]

7. Anti-IL-17

395

Busse and colleagues described results about a clinical trial of phaseIIawith in asthmatic patients with theanti-IL-17 MAb Brodalumab at different doses. The safety was evaluated mon-itoring adverse events and laboratory parameters. The occur-rence of AEs was similar for all dosages, and the most

400

common events were described in upper and lower airways (asthma, upper respiratory tract infection, nasopharyngitis, oral candidiasis, and sinusitis), local injection site reaction,

and cutaneous eventssuch as erythema. AEs were most fre-quent in the brodalumab group than in placebo one,

includ-405

ing events that led to discontinuation. During the trial, seven patients showed serious adverse events, and two patients (one active and one placebo) had an asymptomatic decrease of neutrophil count.[71]

8. TNF-α antagonists

410

Among the TH2-low or no primarilyTH2-driven asthma some phenotypes are well recognized including: adult onset and obesity associated asthma and overlapping syndromes.[72– 74] In such cases,a neutrophilic or pauci-cellular, rather than eosinophilic, inflammation seems to predominate. Based on

415

these observations,a mainly TH1-driven inflammation strategy was hypothesized. TNF-α is known as a central mediator of the

Th1 inflammation, involved in the pathogenesis of many inflammatory diseases, such as Crohn disease, rheumatoid

(9)

arthritis, and various neuropathies.[75,76] In some forms of

420 asthma, TNF-α can enhance mucus production, smooth

mus-cle proliferation, and epithelial disruption. Thus, TNF-α

antag-onism was hypothesized to be an optimal therapeutic target. There were some already in use agents (TNF-α soluble

recep-tor, etanercept, or TNF-α antagonists golimumab and inflixi-425 mab) to bealsotestedin asthma. The earliest studies showed some beneficial effect of etanercept and infliximab in refrac-tory asthma where increased levels of TNF-α were well

ascer-tained.[77–79] Nonetheless, similar studies performed in COPD evidenced an excess of SAEs,such as malignancies,[80] and a

430 randomized double-blind placebo controlled trial in asthma had to be interrupted due to the unacceptable rate of AEs.[81] However, these data must be compared with those obtained in anti-TNF-α-treated patients suffering from rheumatic dis-eases in which there was no evidence for an increased risk

435 of lymphoma and solid cancers.[82–84] In addition, those diagnosed with cancer during or after anti- TNF-α treatment

have not been shown to have a worse prognosis than anti-TNF-α naive patients.[85] Noticeably, the frequency of local (injection site) reactions with TNF-α antagonists is overall low 440 (5%).[86]

Although the TNF-α antagonism remains a reasonable

strategy in some forms of asthma, the available results in this kind of patients display an unfavorable benefit-to-risk rate, and this approach in asthma has been abandoned.

445 9. Conclusions

The knowledge on the pathogenic mechanisms of asthma has rapidly evolved in the past two decades. Although a precise phenotyping or endotyping cannot still be well defined, some biological characteristics can be reasonably attributed to some

450 specific forms of asthma (e.g. TH-2 high or TH2-low). In this context, biological drugs (namely monoclonal antibodies) spe-cifically constructed to antagonize relevant mediators and cytokines (e.g. IgE, IL-5, IL-4, IL-13) have been clinically tested. According to the available results, many oftheseapproaches

455 are clinically effective especially in the most severe forms of asthma. In parallel, the safety aspects of biological drugs became emergent. So far, the safety profile resultsfavorable

for anti-IgE and anti-IL-5 monoclonal antibodies, which are undergoing clinical trials and practical use since many years.

460

The efficacy/safety ratio remains a matter of debate for the new biologicals (anti-IL-13,anti-IL-4,andanti-IL-17), which are more recent in development (Table 3). Some other approaches, that are justified from an immunological point of view (TNF-α antagonism), produced an unacceptable rate

465

of severe adverse events, and were abandoned at least in asthma.

In the near future, the‘personalized’ medical approach will become more and more relevant,[87] with an appropriate investigation of biomarkers. Nonetheless, it must be kept in

470

mind that acting on immunological effectors is not totally devoid of risk,[11] and that a careful and strict surveillance is mandatory needed.

10. Expert opinion

Asthma is currently recognized as a heterogeneous disease,

475

and different phenotypes/endotypes have been proposed in thepast decade. This fact is directly linked to the more and more detailed knowledge on the mechanisms underlying the inflammatory aspects (mainly derived from the allergic model). In this framework, many sensible molecular targets have been

480

identified, which can be selectively blocked by monoclonal antibodies (biological agents). An anti-IgE treatment is already in clinical use since more than 10 years, whereas other biolo-gical are under investigation in clinical trials, often with good clinical results. The increasing number of patients treated,

485

both in clinical practice and trials, attracted the medical atten-tion on the safety aspect, especiallyfor those drugs that are expected to be commercialized soon.

Indeed, the field is of primary relevance, since all those biological agents are used for severe/refractory/steroi

d-490 dependent asthma, which represent a clinical challenge and a socioeconomic burden, and where the side effects of sys-temic steroids are a real occurrence. The results so far available are encouraging in term of clinical efficacy, and the safety is overall satisfactory. The majority of side effects reported are

495

nonspecific (e.g. headache, flu-like symptoms, local reactions) and mild, but treatment-emergent severe adverse events (e.g. anaphylaxis with anti-IgE) can exceptionally occur. Some events seem to be drug-specific (e.g. eosinophilia with dupi-lumab or anti-IL-13), but difficult to interpret from a

patho-500

genic viewpoint. So far, no clear risk factor for severe adverse

Table 3.Brief summary of the reported adverse events in clinical trials (percentage of patients are in parentheses).

Treatment Common AEs Special or severe AEs

Anti-IgE (omalizumab, Xolair™)

Nasopharyngitis, headache, induration at injection site (<10%)

Anaphylaxis-like reactions (~0.09%)

Churg–Strauss syndrome(exceptional case reports) Anti IL-5 (mepolizumab,

Nucala™)

Nasopharyngitis, headache, upper respiratory infection (10–25% of patients. Injection site reaction (<10% of patients)

Anti-mepolizumabAb detection (5%)

Onecase of anaphylaxis with s.c. administration Anti IL-5 (reslizumab) Similar occurrence in placebo and active groups Twocases of anaphylaxis (0.05%) with i.v. administration Anti IL-4/IL-13 (dupilumab) Pharyngitis, headache (8–13%) Injection site reaction

(30%)

Onecase of angioedema

4/52 (7%) hypereosinophilia in the active group Anti IL-13 (lebrikizumab) Injection site reactions (~10%)

Similar rate of AEs in active and placebo groups, mostly mild

Onecase of Lofgren syndrome and 1 case of low platelet count probably treatmentrelated

Anti IL-13 (tralokinumab) Asthma, headache, and nasopharyngitis more frequent in active group (6–13%)

Urinary tract infection (4%) Increased eosinophils (2–6%) 6 G. PASSALACQUA ET AL.

(10)

events could be identified in clinical trials, but this should induce caution in view of thecommercialization, since some events are quite rare and their pathogenesis can be identified only in the post-marketing phase, as happened for alpha-gal

505 and cetuximab.[88] Based on the previous experience with anti-IgE, it is opinion of the authors that a precautional period of observation (at least 30 minafter administration) should be recommended for the newly marketed treatments. Another weak aspect of MAb-based treatment in asthma is that the

510 effects of most cytokines are extensively overlapping, so that the blockage of a single molecule may be insufficient to achieve a meaningful effect.

Within this context, it is clear that the investigation on the role of MAbs in asthma therapy remains of primary relevance

515 due to the fact that it covers those forms of asthma where standard therapy is not sufficient. On the other hand, this approach will not be fully effective until specific biomarkers to identify the subjects suitable for a given treatment and who will respond to that treatment are defined. Currently, very few

520 biomarkers with respect to the number of possible targets and drugsare indeed available (e.g. IgE, bronchial/circulating eosi-nophils, and periostin). This is the optimal goal of which is called‘precision’ or ‘personalized’ medicine.

The field of research with MAbs for ‘difficult asthma’ will

525 certainly remain alive and progressing in the near future, also because the basic research on the mechanisms will be further refined. In addition, it is expected that the development of technology will lead to a sensible reduction in the costs of biological, that remains a main drawback. In the very near

530 future, several anti-IL-5 compounds will be commercialized based on the good results obtained in clinical trials. This biological represents a special case, since IL-5 is specifically committed to eosinophils and has relatively few actions over-lapping with other cytokines. In fact, clinical results are

rele-535 vant in those patients with high eosinophil count (that represent in turn the biomarker). Another expected entry in our armamentarium would be dupilumab, since it antagonizes both IL-4 and IL-13 at the same time, and thus can partially overcome the overlapping effects of the two cytokines. The

540 commercial use ofanti-IL-4 or IL-13 alone,anti-IL-17 seemsto be a more distant perspective.

It is clearthat when a new drug is released for clinical use, the post-marketing data on the safety start to rapidly accu-mulate, and this is expected to be advantageous to identify

545 the possible risk factor and to better define the safety to efficacy ratio.

Declaration of interests

This work was partly supported by ARMIA (Associazione Ricerca Malattie Immunologiche e Allergiche). The authors have no other relevant

affilia-550 tions or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materi-als discussed in the manuscript apart from those disclosed.

ORCID

Andrea Matucci http://orcid.org/0000-0002-8713-4161

555 References

Papers of special note have been highlighted as: • of interest

•• of considerable interest

1. Rosenberg SR, Kalhan R, Mannino DM. Epidemiology of chronic

560

obstructive pulmonary disease: prevalence, morbidity, mortal-ity, and risk factors. SeminRespirCrit Care Med. 2015;36:457–

469. AQ9

2. Accordini S, Corsico AG, Calciano L, et al. The impact of asthma, chronic bronchitis and allergic rhinitis on all-cause hospitalizations

565

and limitations in daily activities: a population-based observational study. BMC Pulm Med.2015;15:10.

3. Global Initiative for the management of Asthma.2015; cited 2016

Feb.www.ginasthma.org

4. Agache I, Akdis C, Jutel M, et al. Untangling asthma phenotypes

570

and endotypes. Allergy.2012;67:835–846.

5. Lötvall J, Akdis CA, Bacharier LB, et al. Asthma endotypes: a new approach to classification of disease entities within the asthma syndrome. J Allergy ClinImmunol.2011;127:355–360.

6. Hambly N, Nair P. Monoclonal antibodies for the treatment of

575

refractory asthma. CurrOpinPulm Med.2014;20:87–89.

7. Chung KF. New treatments for severe treatment-resistant asthma: targeting the right patient. Lancet Respir Med.2013;8:639–652. •• An up-to-date and complete review on the pathogenic

mechanisms of asthma and the use of monoclonal antibodies.

580

8. Canonica GW, Passalacqua G. AIT (allergen immunotherapy): a model for the“precision medicine”. Clin Mol Allergy.2015;13:24. 9. Hamburg MA, Collins FS. The path to personalized medicine. N

Engl J Med.2010;363:301–304.

• The definition and implications of precision medicine that

585

should be applied to the clinical use of monoclonal antibodies. 10. Pelaia G, Vatrella A, Maselli R. The potential of biologics for the

treatment of asthma. Nat Rev Drug Discov.2012;11:958–972. 11. Singh JA, Wells GA, Christensen R, et al. Adverse effects of

biolo-gics: a network meta-analysis and cochrane overview. Cochrane

590

Database Syst Rev. 2011;;CD008794. AQ10

•• An interesting and advanced meta-analysis, showing that monoclonal antibodies for different diseases carry some risk of adverse events.

12. Hansel TT, Kropshofer H, Singer T, et al. The safety and side effects

595

of monoclonal antibodies. Nat Rev Drug Discov.2010;9:325–338. 13. Cherwinski HM, Schumacher JH, Brown KD, et al. Two types of

mouse helper T cell clone. III. Further differences in lymphokine synthesis between Th1 and Th2 clones revealed by RNA hybridiza-tion, functionally monospecific bioassays, and monoclonal

antibo-600

dies. J Exp Med.1986;166:1229.

14. Del Prete GF, De Carli M, Mastromauro C, et al. Purified protein derivative (PPD) of Mycobacterium tuberculosis and excretory-secretory antigen(s) (TES) of Toxocaracanis select human T cells clones with stable and opposite (Th1 or Th2) profile of cytokine

605

production. J Clin Invest.1991;88:346–350.

15. Halim TY, Krauss RH, Sun AC, et al. Lung natural helper cells are a critical source of TH2 cell-type cytokines in protease allergen-induced airway inflammation. Immunity.2012;36:451–463. 16. Takayama G, Arima K, Kanaji T, et al. Periostin: a novel component

610

of subepithelial fibrosis of bronchial asthma downstream of IL-4 and IL-13 signals. J Allergy Clin Immunol.2006;118:98–104. 17. Humbert M, Durham SR, Ying S. IL-4 and IL-5 mRNA and protein in

bronchial biopsies from patients with atopic and non-atopic asthma: evidence against“intrinsic” asthma being a distinct

immu-615

nopathologic entity. Am J Respir Crit Care Med.1996;154:1497– 1504.

18. Zoratti E, Havstad S, Wegienka G, et al. Johnson CCDifferentiating asthma phenotypes in young adults through polyclonal cytokine profiles. Ann Allergy Asthma Immunol.2014;113:25–30.

620

19. Lee JJ, Jacobsen EA, McGarry MP, et al. Eosinophils in health and disease: the LIAR hypothesis. Clin ExpAllergy.2010;40:563–575. 20. Ray A, Oriss TB, Wenzel SE. Emerging molecular phenotypes of

asthma. Am J Physiol Lung Cell Mol Physiol.2015;308:L130–40. EXPERT OPINION ON DRUG SAFETY 7

(11)

21. Shen ZJ, Malter JS. Determinants of eosinophil survival and

apop-625 totic cell death. Apoptosis.2015;20:224–234.

22. Smith SG, Hill M, Oliveria JP, et al. Evaluation of peroxisome pro-liferator-activated receptor agonists on interleukin-5-induced eosi-nophil differentiation. Immunology.2014;142:484–491.

23. Ravensberg AJ, Fml R, Van Schadewijk A, et al. Eotaxin-2 and

630 eotaxin-3 expression is associated with persistent eosinophilic bronchial inflammation in patients with asthma after allergen chal-lenge. J Allergy ClinImmunol.2005;115:779–785.

24. Varricchi G, Bagnasco D, Borriello F, et al. Interleukin-5 pathway inhibition in the treatment of eosinophilic respiratory disorders:

635 evidence and unmet needs. Curr Opin Allergy ClinImmunol. 2016;16:186–200.

• A review on the biological effects of IL-5 in asthma and its potential role as main therapeutic target.

25. Schuijs MJ, Willart MA, Hammad H. Cytokine targets in airway

640 inflammation. Curr Opinion Pharmacol.2013;13:351–361. 26. Lambrecht BN, Hammad H. The immunology of asthma. Nat

Immunol.2015;16:45–56.

27. Maggi E, Vultaggio A, Matucci A. Acute infusion reactions induced by monoclonal antibody therapy. Expert Rev ClinImmunol.2011;7

645 (1):55–63.

28. Vultaggio A, Castells MC. Hypersensitivity reactions to biologic agents. Immunol Allergy Clin North Am.2014;34:615–632. •• One of the first and most updated review on the possible

reactions to biologicals and their putative mechanisms.

650 29. Proctor L, Renzulli B, Warren S, et al. Monoclonal antibody-stimu-lated serum sickness. Transfusion.2004;44(7):955.

30. Matucci A, Maggi E, Vultaggio A. Cellular and humoral immune responses during tuberculosis infection: useful knowledge in the era of biological agents. J Rheumatol Suppl.2014;91:17–23.

655 31. Busse W, Corren J, Lanier BQ, et al. Omalizumab, anti-IgE recombi-nant humanized monoclonal antibody, for the treatment of severe allergic asthma. J Allergy ClinImmunol.2001;108:184–190. 32. Casale TB, Condemi J, LaForce C, et al. Omalizumab seasonal

allergic rhinitis trail group. Effect of Omalizumab on Symptoms of

660 Seasonal Allergic Rhinitis: a Randomized Controlled Trial JAMA. 2001;286:2956–2967.

33. Lai T, Wang S, Xu Z, et al. Long-term efficacy and safety of omali-zumab in patients with persistent uncontrolled allergic asthma: a systematic review and meta-analysis. Sci Rep.2015;5:8191.

665 34. Tsabouri S, Tseretopoulou X, Priftis K, et al. Omalizumab for the treatment of inadequately controlled allergic rhinitis: a systematic review and meta-analysis of randomized clinical trials. J Allergy ClinImmunolPract.2014;2:332–340.

35. Normansell R, Walker S, Milan SJ, et al. Omalizumab for asthma in

670 adults and children. Cochrane Database Syst Rev. 2014;1; CD003559.

AQ11

36. Tan RA, Corren J. Safety of omalizumab in asthma. Expert Opin Drug Saf.2011;10:463–471.

37. Corren J, Casale TB, Lanier B, et al. Safety and tolerability of

675 omalizumab. ClinExp Allergy.2009;39:788–797.

38. Long A, Rahmaoui A, Rothman KJ, et al. Incidence of malignancy in patients with moderate-to-severe asthma treated with or without omalizumab. J Allergy Clin Immunol.2014;134:560–567.

39. Busse W, Buhl R, Fernandez Vidaurre C, et al. Omalizumab and the

680 risk of malignancy: results from a pooled analysis. J Allergy Clin Immunol.2012;129:983–989.

40. Cooper PJ, Ayre G, Martin C, et al. Geohelminth infections: a review of the role of IgE and assessment of potential risks of anti-IgE treatment. Allergy.2008;63:409–417.

685 41. Cruz AA, Lima F, Sarinho E, et al. Safety of anti-immunoglobulin E therapy with omalizumab in allergic patients at risk of geohelminth infection. ClinExpAllergy.2007;37:197–207.

42. Jandus P, Hausmann O, Haeberli G, et al. Unpredicted adverse reaction to omalizumab. J Investig Allergo Lclin Immunol.

690 2011;21:563–566.

43. Lin RY, Rodriguez-Baez G, Bhargave GA. Omalizumab-associated anaphylactic reactions reported between January 2007 and June 2008. Ann Allergy Asthma Immunol.2009;103:442–445.

44. Price KS, Hamilton RG. Anaphylactoid reactions in two patients

695

after omalizumab administration after successful long-term ther-apy. AllergyAsthma Proc.2007;28:313–319.

45. Cox L, Lieberman P, Wallace D, et al. American Academy of Allergy, Asthma & Immunology/American College of Allergy, Asthma & Immunology Omalizumab-Associated Anaphylaxis Joint Task

700

Force follow-up report. J Allergy Clin Immunol.2011;128:210–212. • The description and evaluation of anaphylaxis induced by omalizumab performed in a large population over many years. 46. El-Qutob D. Off-label uses of Omalizumab. Clin Rev Allergy

Immunol.2016;50:84–96.

705

47. Gleich JG. Mechanisms of eosinophil-associated inflammation. J All Clin Immunol.2000;105:651–663.

48. Kolbeck R, Kozhich A, Koike M, et al. MEDI-563, a humanized anti-IL-5 receptor alpha mAb with enhanced antibody-dependent cell-mediated cytotoxicity function. J Allergy Clin Immunol.

710

2010;125:1344–1353.

49. Busse WW, Katial R, Gossage D, et al. Safety profile, pharmacoki-netics, and biologic activity of MEDI-563, an anti-IL-5 receptor alpha antibody, in a phase I study of subjects with mild asthma. J Allergy Clin Immunol.2010;125:1237–1244.

715

50. Laviolette M, Gossage DL, Gauvreau G, et al. Effects of benralizu-mab on airway eosinophils in asthmatic patients with sputum eosinophilia. J Allergy Clin Immunol.2013;132:1086–1096. 51. Castro M, Wenzel SE, Bleecker ER, et al. Benralizumab, an

anti-interleukin 5 receptorα monoclonal antibody, versus placebo for

720

uncontrolled eosinophilic asthma: a phase 2b randomised dose-ranging study. Lancet Respir Med.2014;2:879–890.

52. Haldar P, Brightling CE, Hargadon B, et al. Mepolizumab and exacerbations of refractory eosinophilic asthma. N Engl J Med. 2009;360:973–984.

725

53. Nair P, Pizzichini MM, Kjarsgaard M, et al. Mepolizumab for pre-dnisone-dependent asthma with sputum eosinophilia. N Engl J Med.2009;360:985–993.

54. Pouliquen I, Kornmann O, Barton SV, et al. Characterization of the relationship between dose and blood eosinophil response

follow-730

ing subcutaneous administration of mepolizumab. Int J Clin Pharmacol Therapeutics.2015;53:1015–1027.

55. Ortega HG, Liu MC, Pavord ID, et al. Mepolizumab treatment in patients with severe eosinophilic asthma. N Engl J Med. 2014;371:1198–1207.

735

56. Pavord ID, Korn S, Howarth P, et al. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, pla-cebo-controlled trial. Lancet.2012;380:651–659.

57. Liu Y, Zhang S, Li DW, et al. Efficacy of anti-interleukin-5 therapy with mepolizumab in patients with asthma: a meta-analysis of

740

randomized placebo-controlled trials. PLoS One. 2013;8(3): e59872.

58. Castro M, Mathur S, Hargreave F, et al. Reslizumab for poorly controlled, eosinophilic asthma: a randomized, placebo-controlled study. Am J Respir Crit Care Med.2011;184:1125–1132.

745

59. Castro M, Zangrilli J, Wechsler ME, et al. Reslizumab for inade-quately controlled asthma with elevated blood eosinophil counts: results from two multicentre, parallel, double-blind, randomised, placebo-controlled, phase 3 trials. Lancet Respir Med. 2015;3 (5):355–366.

750

60. Herndon FJ, Kayes SG. Depletion of eosinophils by anti-IL-5 mono-clonal antibody treatment of mice infected with Trichinella spiralis does not alter parasite burden or immunologic resistance to rein-fection. J Immunol.1992;149:3642–3647.

61. Geich GJ, Klion AD, Lee JJ, et al. The consequences of not having

755

eosinophils. Allergy.2013;68:829–835.

62. Sakkal S, Miller S, Apostolopoulos V, et al. Eosinophils in cancer:

favourable or unfavourable? Curr Med Chem.2016. AQ12 63. Roufosse FE, Kahn JE, Gleich GJ, et al. Long-term safety of

mepo-lizumab for the treatment of hypereosinophilic syndromes. J

760

Allergy Clin Immunol.2013;131:461–467.

64. Wenzel S, Ford L, Pearlman D, et al. Dupilumab in persistent asthma with elevated eosinophil levels. N Engl J Med. 2013;368:2455–2466.

(12)

65. Wenzel S, Wilbraham D, Fuller R, et al. Effect of an interleukin-4

765 variant on late phase asthmatic response to allergen challenge in asthmatic patients: results of two phase 2a studies. Lancet. 2007;370:1422–1431.

• An interesting summary of the efficacy and safety of IL-4 antag-onism (pitrakinra) in the only available phase II clinical trials.

770 66. Hanania A, Noonan M, Corren J, et al. Lebrikizumab in moderate-to-severe asthma: pooled data from two randomised placebo-con-trolled studies. Thorax.2015;70:748–756.

67. Corren J, Lemanske RF, Hanania NA, et al. Lebrikizumab treatment in adults with asthma. N Engl J Med.2011;365:1088–1098.

775 68. Noonan M, Korenblat P, Mosesova S, et al. Dose-ranging study of lebrikizumab in asthmatic patients not receiving inhaled steroids. J Allergy Clin Immunol.2013;132:567–574.

69. Scheerens H, Arron JR, Zheng Y, et al. The effects of lebrikizumab in patients with mild asthma following whole lung allergen challenge.

780 Clin Exp Allergy.2014;44:38–46.

70. Piper E, Brightling C, Niven R, et al. A phase II placebo-controlled study of tralokinumab in moderate-to-severe asthma. Eur Respir J. 2013;41:330–338.

71. Busse WW, Holgate S, Kerwin E, et al. Randomized, double-blind,

785 placebo-controlled study of brodalumab, a human anti-IL-17 recep-tor monoclonal antibody, in moderate to severe asthma. Am J Respir Crit Care Med.2013;188:1294–1302.

• The first clinical trial with anti-IL-17, showing indeed only marginal results.

790 72. Ding B, Enstone A. Asthma and chronic obstructive pulmonary disease overlap syndrome (ACOS): structured literature review and physician insights. Expert Rev Respir Med.2016;15:1–9. 73. Alshabanat A, Zafari Z, Albanyan O, et al. Asthma and COPD

over-lap syndrome (ACOS): a systematic review and meta-analysis.

795 PLoSOne.2015;10:0136065.

74. Leiria LO, Martins MA, Saad MJ. Obesity and asthma: beyond T(H)2 inflammation. Metabolism.2015;64:172–181.

75. Torres J, Boyapati RK, Kennedy NA, et al. Systematic review of effects of withdrawal of immunomodulators or biologic agents

800 from patients with inflammatory bowel disease. Gastroenterology. 2015;49:1716–1730.

76. Stübgen JP. Tumor necrosis factor-alpha as a potential therapeutic target in idiopathic inflammatory myopathies. J Neurol. 2011;258:961–970.

805

77. Morjaria JB, Chauhan AJ, Babu KS, et al. The role of a soluble TNFalpha receptor fusion protein (etanercept) in corticosteroid refractory asthma: a double blind, randomised, placebo controlled trial. Thorax.2008;63:584–591.

78. Berry MA, Hargadon B, Shelley M, et al. Evidence of a role of tumor

810

necrosis factor alpha in refractory asthma. N Engl J Med. 2006;354:697–708.

79. Holgate ST, Noonan M, Chanez P, et al. Efficacy and safety of etanercept in moderate-to-severe asthma: a randomised, con-trolled trial. Eur Respir J.2011;37:1352–1359.

815

80. Brightling C, Berry M. Amrani Y targeting TNF-alpha: a novel ther-apeutic approach for asthma. J Allergy Clin Immunol.2008;121:5–10. 81. Wenzel SE, Barnes PJ, Bleecker ER, et al. Arandomized, double-blind, placebo-controlledstudy of tumornecrosisfactor-alphablock-ade in severe persistentasthma. AmJ Respir Crit Care Med.

820

2009;179:549–558.

82. Askling J, Fored CM, Brandt L, et al. Risks of solid cancers in patients with rheumatoid arthritis and after treatment with tumour necrosis factor antagonists. Annals of the Rheumatic Diseases. 2005;64:1421–1426.

825

83. Strangfeld A, Hierse F, Rau R, et al. Risk of incident or recurrent malignancies among patients with rheumatoid arthritis exposed to biologic therapy in the German biologics register RABBIT. Arthritis Research & Therapy.2010;12(1):R5.

84. Carmona L, Abasolo L, Descalzo MA, et al. Cancer in patients with

830

rheumatic diseases exposed to TNF antagonists. Semin Arthritis Rheum2011;41:71–80.

85. Raaschou P, Simard JF, Neovius M, et al. Does cancer that occurs during or after anti-tumor necrosis factor therapy have a worse prognosis? A national assessment of overall and site-specific cancer

835

survival in rheumatoid arthritis patients treated with biologic agents. Arthritis and Rheumatism.2011;63:1812–1822.

86. Vultaggio A, Matucci A, Nencini F, et al. Anti-infliximab IgE and non IgE antibodies and induction of infusion-related severe anaphylac-tic reactions. Allergy.2010;65:657–661.

840

87. De Ferrari L, Chiappori A, Bagnasco D, et al. Molecular phenotyping and biomarker development: are we on our way towards targeted therapy for severe asthma? Expert Rev Respir Med.2016;10:29–38. 88. Steinke JW, Platts-Mills TA, Commins SP. The alpha-gal story: les-sons learned from connecting the dots. J Allergy Clin Immunol.

845

2015;135:589–596.

Riferimenti

Documenti correlati

Dopo aver esaminato le principali indicazioni della prassi e della dottrina in tema di valutazione dei beni culturali, gli autori affrontano un caso di studio nel quale è

diversi. L’opera è il frutto della pluridecennale atti- vità di ricerca, tutela e valorizzazione che il curato- re ha dedicato alla città e al suo territorio; gli autori

N pts: number of patients; SMM: smoldering multiple myeloma; Dara: Daratumumab; CR: complete response; PFS: progression-free survival; NDMM: newly diagnosed multiple myeloma;

In the light of this, it has been decided to try a different kind of selection, the main goal was to find a protocol that allowed the isolation from the naïve phage display library

Il pathway alternativo richiede (per la sintesi di purine e thymidylate) l’apporto esogeno di hypoxanthine e thymidine, che infatti sono insieme all’ aminopterin i componenti del

In conclusion we found that subsets of Arg residues at specific locations in the CDRs of heavy and light chains of pathogenic aPL are important in determining their ability to bind CL

The addition of rituximab to a combination of fludarabine, cyclophosphamide, mitoxantrone (FCM) significantly increases the response rate and prolongs survival as compared with

Based on the observed clinical efficacy and the medical need in the treatment of patients with MS, the Committee for Medicinal Products for Human Use (CHMP, until 2004: CPMP) at